Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 281
Filter
1.
Ren Fail ; 46(1): 2349139, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38712768

ABSTRACT

BACKGROUND: NOP2/Sun RNA methyltransferase 5 (NSUN5) is an RNA methyltransferase that has a broad distribution and plays critical roles in various biological processes. However, our knowledge of the biological functions of NSUN5 in mammals is very limited. Therefore, in this study, we investigate the role of NSUN5 in mice. METHODS: In the present research, we built a mouse model (Nsun5-/-) using the CRISPR/Cas9 system to investigated the specific role of NSUN5. RESULTS: We observed that Nsun5-/- mice had a reduced body weight compared to wild-type mice. Additionally, their survival rate gradually decreased to 20% after postnatal day (PD) 21. Further examination revealed the Nsun5-/- mice had multiple organ damage, with the most severe damage occurring in the kidneys. Moreover, we observed glycogen deposition and fibrosis, along with a notable shorting of the primary foot processes of glomeruli in Nsun5-/- kidneys. Furthermore, we found that the kidneys of Nsun5-/- mice showed increased expression of the apoptotic signal Caspase-3 and accumulated stronger DNA damage at PD 21. CONCLUSIONS: In our study, we found that mice lacking NSUN5 died before puberty due to kidney fatal damage caused by DNA damage and cell apoptosis. These results suggest that NSUN5 plays a vital role in preventing the accumulation of DNA damage and cell apoptosis in the kidney.


Subject(s)
Apoptosis , Kidney , Methyltransferases , Mice, Knockout , Animals , Mice , Methyltransferases/genetics , Methyltransferases/metabolism , Methyltransferases/deficiency , Kidney/pathology , Disease Models, Animal , DNA Damage , Kidney Diseases/genetics , Kidney Diseases/pathology , Male , Mice, Inbred C57BL , CRISPR-Cas Systems , Caspase 3/metabolism
2.
Nucleic Acids Res ; 51(9): 4341-4362, 2023 05 22.
Article in English | MEDLINE | ID: mdl-36928661

ABSTRACT

BRCA1 mutations are associated with increased breast and ovarian cancer risk. BRCA1-mutant tumors are high-grade, recurrent, and often become resistant to standard therapies. Herein, we performed a targeted CRISPR-Cas9 screen and identified MEPCE, a methylphosphate capping enzyme, as a synthetic lethal interactor of BRCA1. Mechanistically, we demonstrate that depletion of MEPCE in a BRCA1-deficient setting led to dysregulated RNA polymerase II (RNAPII) promoter-proximal pausing, R-loop accumulation, and replication stress, contributing to transcription-replication collisions. These collisions compromise genomic integrity resulting in loss of viability of BRCA1-deficient cells. We also extend these findings to another RNAPII-regulating factor, PAF1. This study identifies a new class of synthetic lethal partners of BRCA1 that exploit the RNAPII pausing regulation and highlight the untapped potential of transcription-replication collision-inducing factors as unique potential therapeutic targets for treating cancers associated with BRCA1 mutations.


Subject(s)
BRCA1 Protein , DNA Replication , Hereditary Breast and Ovarian Cancer Syndrome , Mutation , Transcription, Genetic , Humans , BRCA1 Protein/deficiency , BRCA1 Protein/genetics , DNA Replication/genetics , Hereditary Breast and Ovarian Cancer Syndrome/genetics , Hereditary Breast and Ovarian Cancer Syndrome/pathology , Hereditary Breast and Ovarian Cancer Syndrome/physiopathology , RNA Polymerase II/metabolism , Transcription, Genetic/genetics , Promoter Regions, Genetic , Methyltransferases/deficiency , Methyltransferases/genetics , R-Loop Structures , Cell Death
3.
Mol Ther ; 30(12): 3714-3728, 2022 12 07.
Article in English | MEDLINE | ID: mdl-35923112

ABSTRACT

Activation of hepatic stellate cells (HSCs) is a central driver of liver fibrosis. Previous investigations have identified various altered epigenetic landscapes during the cellular progression of HSC activation. N6-methyladenosine (m6A) is the most abundant internal RNA modification in eukaryotic cells and is dynamically regulated under various physiological and pathophysiological conditions. However, the functional role of Mettl3-mediated m6A in liver fibrosis remains elusive. Here, we found that the HSC-specific knockout of m6A methyltransferase Mettl3 suppressed HSC activation and significantly alleviated liver fibrosis. Multi-omics analysis of HSCs showed that Mettl3 depletion reduced m6A deposition on mRNA transcripts of Lats2 (a central player of the Hippo/YAP signaling pathway) and slowed down their degradation. Elevated Lats2 increased phosphorylation of the downstream transcription factor YAP, suppressed YAP nuclear translocation, and decreased pro-fibrotic gene expression. Overexpressing YAP mutant resistant to phosphorylation by Lats2 partially rescued the activation and pro-fibrotic gene expression of Mettl3-deficient HSCs. Our study revealed that disruption of Mettl3 in HSCs mitigated liver fibrosis by controlling the Hippo/YAP signaling pathway, providing potential therapeutic strategies to alleviate liver fibrosis by targeting epitranscriptomic machinery.


Subject(s)
Hepatic Stellate Cells , Liver Cirrhosis , Methyltransferases , Liver Cirrhosis/genetics , Methyltransferases/deficiency , Methyltransferases/genetics , Multiomics , Protein Serine-Threonine Kinases/genetics , Tumor Suppressor Proteins , Animals , Mice
4.
J Biol Chem ; 298(3): 101692, 2022 03.
Article in English | MEDLINE | ID: mdl-35148993

ABSTRACT

We previously reported that loss of mitochondrial transcription factor B1 (TFB1M) leads to mitochondrial dysfunction and is involved in the pathogenesis of type 2 diabetes (T2D). Whether defects in ribosomal processing impact mitochondrial function and could play a pathogenetic role in ß-cells and T2D is not known. To this end, we explored expression and the functional role of dimethyladenosine transferase 1 homolog (DIMT1), a homolog of TFB1M and a ribosomal RNA (rRNA) methyltransferase implicated in the control of rRNA. Expression of DIMT1 was increased in human islets from T2D donors and correlated positively with expression of insulin mRNA, but negatively with insulin secretion. We show that silencing of DIMT1 in insulin-secreting cells impacted mitochondrial function, leading to lower expression of mitochondrial OXPHOS proteins, reduced oxygen consumption rate, dissipated mitochondrial membrane potential, and a slower rate of ATP production. In addition, the rate of protein synthesis was retarded upon DIMT1 deficiency. Consequently, we found that DIMT1 deficiency led to perturbed insulin secretion in rodent cell lines and islets, as well as in a human ß-cell line. We observed defects in rRNA processing and reduced interactions between NIN1 (RPN12) binding protein 1 homolog (NOB-1) and pescadillo ribosomal biogenesis factor 1 (PES-1), critical ribosomal subunit RNA proteins, the dysfunction of which may play a part in disturbing protein synthesis in ß-cells. In conclusion, DIMT1 deficiency perturbs protein synthesis, resulting in mitochondrial dysfunction and disrupted insulin secretion, both potential pathogenetic processes in T2D.


Subject(s)
Diabetes Mellitus, Type 2 , Insulin-Secreting Cells , Methyltransferases , Mitochondria , Ribosomes , Animals , Diabetes Mellitus, Type 2/metabolism , Humans , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/metabolism , Methyltransferases/deficiency , Methyltransferases/metabolism , Mitochondria/metabolism , RNA, Ribosomal/genetics , RNA, Ribosomal/metabolism , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism , Ribosomes/metabolism , Transferases/metabolism
5.
Nat Commun ; 13(1): 168, 2022 01 10.
Article in English | MEDLINE | ID: mdl-35013323

ABSTRACT

Skeletal muscle serves fundamental roles in organismal health. Gene expression fluctuations are critical for muscle homeostasis and the response to environmental insults. Yet, little is known about post-transcriptional mechanisms regulating such fluctuations while impacting muscle proteome. Here we report genome-wide analysis of mRNA methyladenosine (m6A) dynamics of skeletal muscle hypertrophic growth following overload-induced stress. We show that increases in METTL3 (the m6A enzyme), and concomitantly m6A, control skeletal muscle size during hypertrophy; exogenous delivery of METTL3 induces skeletal muscle growth, even without external triggers. We also show that METTL3 represses activin type 2 A receptors (ACVR2A) synthesis, blunting activation of anti-hypertrophic signaling. Notably, myofiber-specific conditional genetic deletion of METTL3 caused spontaneous muscle wasting over time and abrogated overload-induced hypertrophy; a phenotype reverted by co-administration of a myostatin inhibitor. These studies identify a previously unrecognized post-transcriptional mechanism promoting the hypertrophic response of skeletal muscle via control of myostatin signaling.


Subject(s)
Activin Receptors, Type II/genetics , Hypertrophy/genetics , Methyltransferases/genetics , Muscle, Skeletal/metabolism , Muscular Atrophy/genetics , Myostatin/genetics , Activin Receptors, Type II/metabolism , Adenosine/analogs & derivatives , Adenosine/metabolism , Animals , Dependovirus/genetics , Dependovirus/metabolism , Gene Expression Regulation, Developmental , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Genome-Wide Association Study , Hypertrophy/metabolism , Hypertrophy/pathology , Hypertrophy/prevention & control , Male , Methyltransferases/deficiency , Mice , Muscle Development/genetics , Muscle, Skeletal/pathology , Muscular Atrophy/metabolism , Muscular Atrophy/pathology , Myostatin/metabolism , Signal Transduction
6.
Front Immunol ; 12: 751688, 2021.
Article in English | MEDLINE | ID: mdl-34691066

ABSTRACT

The 5' end of the flavivirus genome contains a type 1 cap structure formed by sequential N-7 and 2'-O methylations by viral methyltransferase (MTase). Cap methylation of flavivirus genome is an essential structural modification to ensure the normal proliferation of the virus. Tembusu virus (TMUV) (genus Flavivirus) is a causative agent of duck egg drop syndrome and has zoonotic potential. Here, we identified the in vitro activity of TMUV MTase and determined the effect of K61-D146-K182-E218 enzymatic tetrad on N-7 and 2'-O methylation. The entire K61-D146-K182-E218 motif is essential for 2'-O MTase activity, whereas N-7 MTase activity requires only D146. To investigate its phenotype, the single point mutation (K61A, D146A, K182A or E218A) was introduced into TMUV replicon (pCMV-Rep-NanoLuc) and TMUV infectious cDNA clone (pACYC-TMUV). K-D-K-E mutations reduced the replication ability of replicon. K61A, K182A and E218A viruses were genetically stable, whereas D146A virus was unstable and reverted to WT virus. Mutant viruses were replication and virulence impaired, showing reduced growth and attenuated cytopathic effects and reduced mortality of duck embryos. Molecular mechanism studies showed that the translation efficiency of mutant viruses was inhibited and a higher host innate immunity was induced. Furthermore, we found that the translation inhibition of MTase-deficient viruses was caused by a defect in N-7 methylation, whereas the absence of 2'-O methylation did not affect viral translation. Taken together, our data validate the debilitating mechanism of MTase-deficient avian flavivirus and reveal an important role for cap-methylation in viral translation, proliferation, and escape from innate immunity.


Subject(s)
Fibroblasts/immunology , Flavivirus/genetics , Methyltransferases/deficiency , RNA, Viral , Animals , Cells, Cultured , Ducks , Embryo, Nonmammalian , Fibroblasts/virology , Immunity, Innate , Mesocricetus , Methylation , Methyltransferases/genetics , Mutation , Viral Proteins/genetics
7.
Nat Commun ; 12(1): 5522, 2021 09 17.
Article in English | MEDLINE | ID: mdl-34535671

ABSTRACT

Natural killer (NK) cells exert critical roles in anti-tumor immunity but how their functions are regulated by epitranscriptional modification (e.g., N6-methyladenosine (m6A) methylation) is unclear. Here we report decreased expression of the m6A "writer" METTL3 in tumor-infiltrating NK cells, and a positive correlation between protein expression levels of METTL3 and effector molecules in NK cells. Deletion of Mettl3 in NK cells alters the homeostasis of NK cells and inhibits NK cell infiltration and function in the tumor microenvironment, leading to accelerated tumor development and shortened survival in mice. The gene encoding SHP-2 is m6A modified, and its protein expression is decreased in METTL3-deficient NK cells. Reduced SHP-2 activity renders NK cells hyporesponsive to IL-15, which is associated with suppressed activation of the AKT and MAPK signaling pathway in METTL3-deficient NK cells. These findings show that m6A methylation safeguards the homeostasis and tumor immunosurveillance function of NK cells.


Subject(s)
Adenosine/analogs & derivatives , Killer Cells, Natural/immunology , Methyltransferases/metabolism , Neoplasms/immunology , Neoplasms/metabolism , RNA/metabolism , Adenosine/metabolism , Animals , Carcinogenesis/pathology , Cell Line, Tumor , Gene Deletion , Homeostasis , Interleukin-15/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Methylation , Methyltransferases/deficiency , Mice, Inbred C57BL , Mice, Knockout , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Tumor Microenvironment
8.
Nat Commun ; 12(1): 4913, 2021 08 13.
Article in English | MEDLINE | ID: mdl-34389722

ABSTRACT

Epitranscriptomic mechanisms linking tRNA function and the brain proteome to cognition and complex behaviors are not well described. Here, we report bi-directional changes in depression-related behaviors after genetic disruption of neuronal tRNA cytosine methylation, including conditional ablation and transgene-derived overexpression of Nsun2 in the mouse prefrontal cortex (PFC). Neuronal Nsun2-deficiency was associated with a decrease in tRNA m5C levels, resulting in deficits in expression of 70% of tRNAGly isodecoders. Altogether, 1488/5820 proteins changed upon neuronal Nsun2-deficiency, in conjunction with glycine codon-specific defects in translational efficiencies. Loss of Gly-rich proteins critical for glutamatergic neurotransmission was associated with impaired synaptic signaling at PFC pyramidal neurons and defective contextual fear memory. Changes in the neuronal translatome were also associated with a 146% increase in glycine biosynthesis. These findings highlight the methylation sensitivity of glycinergic tRNAs in the adult PFC. Furthermore, they link synaptic plasticity and complex behaviors to epitranscriptomic modifications of cognate tRNAs and the proteomic homeostasis associated with specific amino acids.


Subject(s)
Depressive Disorder/physiopathology , Epigenesis, Genetic/genetics , Methyltransferases/genetics , Proteome/metabolism , RNA, Transfer/genetics , Synaptic Transmission/genetics , Animals , Depressive Disorder/genetics , Depressive Disorder/metabolism , Gene Expression Profiling/methods , Methyltransferases/deficiency , Methyltransferases/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neurons/metabolism , Prefrontal Cortex/cytology , Prefrontal Cortex/metabolism , Prefrontal Cortex/physiology , Proteomics/methods , RNA, Transfer/metabolism , Signal Transduction/genetics
9.
Nucleic Acids Res ; 49(9): 5382-5392, 2021 05 21.
Article in English | MEDLINE | ID: mdl-33956156

ABSTRACT

The emergence of SARS-CoV-2 infection has posed unprecedented threat to global public health. The virus-encoded non-structural protein 14 (nsp14) is a bi-functional enzyme consisting of an exoribonuclease (ExoN) domain and a methyltransferase (MTase) domain and plays a pivotal role in viral replication. Here, we report the structure of SARS-CoV-2 nsp14-ExoN domain bound to its co-factor nsp10 and show that, compared to the SARS-CoV nsp10/nsp14-full-length complex, SARS-CoV-2 nsp14-ExoN retains an integral exoribonuclease fold and preserves an active configuration in the catalytic center. Analysis of the nsp10/nsp14-ExoN interface reveals a footprint in nsp10 extensively overlapping with that observed in the nsp10/nsp16 structure. A marked difference in the co-factor when engaging nsp14 and nsp16 lies in helix-α1', which is further experimentally ascertained to be involved in nsp14-binding but not in nsp16-engagement. Finally, we also show that nsp10/nsp14-ExoN is enzymatically active despite the absence of nsp14-MTase domain. These data demonstrate that SARS-CoV-2 nsp10/nsp14-ExoN functions as an exoribonuclease with both structural and functional integrity.


Subject(s)
Biocatalysis , Exoribonucleases/chemistry , Exoribonucleases/metabolism , SARS-CoV-2/chemistry , SARS-CoV-2/enzymology , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/metabolism , Viral Regulatory and Accessory Proteins/chemistry , Viral Regulatory and Accessory Proteins/metabolism , Binding Sites , Crystallography, X-Ray , Exoribonucleases/genetics , Guanine , Methyltransferases/chemistry , Methyltransferases/deficiency , Methyltransferases/genetics , Methyltransferases/metabolism , Models, Molecular , Protein Domains/genetics , SARS-CoV-2/genetics , Viral Nonstructural Proteins/genetics , Viral Regulatory and Accessory Proteins/genetics
10.
Sci Rep ; 11(1): 10965, 2021 05 26.
Article in English | MEDLINE | ID: mdl-34040101

ABSTRACT

DNA methylation plays an important role in modulating plant growth plasticity in response to stress, but mechanisms involved in such control need further investigation. We used drm1 drm2 cmt3 mutant of Arabidopsis thaliana, defective in DNA methylation, to explore metabolic pathways downstream epigenetic modulation under cadmium (Cd) stress. To this aim, a transcriptomic analysis was performed on ddc and WT plants exposed to a long-lasting (21 d) Cd treatment (25/50 µM), focusing on hormone genetic pathways. Growth parameters and hormones amount were also estimated. Transcriptomic data and hormone quantification showed that, under prolonged Cd treatment, level and signalling of growth-sustaining hormones (auxins, CKs, GAs) were enhanced and/or maintained, while a decrease was detected for stress-related hormones (JA, ABA, SA), likely as a strategy to avoid the side effects of their long-lasting activation. Such picture was more effective in ddc than WT, already at 25 µM Cd, in line with its better growth performance. A tight relationship between methylation status and the modulation of hormone genetic pathways under Cd stress was assessed. We propose that the higher genome plasticity conferred to ddc by DNA hypomethylated status underlies its prompt response to modulate hormones genetic pathways and activity and assure a flexible growth.


Subject(s)
Arabidopsis Proteins/physiology , Arabidopsis/genetics , Cadmium/pharmacology , DNA-Cytosine Methylases/physiology , Gene Expression Regulation, Plant/drug effects , Methyltransferases/physiology , Plant Growth Regulators/genetics , Arabidopsis/drug effects , Arabidopsis/growth & development , Arabidopsis Proteins/genetics , Base Sequence , Cytokinins/biosynthesis , Cytokinins/genetics , DNA Methylation , DNA, Plant/genetics , DNA-Cytosine Methylases/deficiency , DNA-Cytosine Methylases/genetics , Genes, Plant , Methyltransferases/deficiency , Methyltransferases/genetics , Mutation , Plant Roots/growth & development , RNA, Messenger/genetics , RNA, Plant/genetics , Soil Pollutants/pharmacology , Stress, Physiological/genetics , Transcriptome/drug effects
11.
Cell Mol Gastroenterol Hepatol ; 12(3): 839-856, 2021.
Article in English | MEDLINE | ID: mdl-33992834

ABSTRACT

BACKGROUND AND AIMS: Transforming growth factor ß1 (TGF-ß1) secreted from activated Kupffer cells (KC) promotes the progression of nonalcoholic steatohepatitis (NASH) to liver fibrosis. N6-methyladenosine (m6A) RNA modification participates in various cell stress responses, yet it remains unknown whether it plays a role in TGF-ß1 upregulation in activated KCs. METHODS: Western blot, dot blot, and liquid chromatography with tandem mass spectrometry were used to determine the expression of m6A methyltransferase, METTL3, and METTL14, as well as global m6A modification. RNA-sequencing and m6A-seq were employed to screen differentially expressed genes and responsive m6A peaks. Nuclear factor κB (NF-κB)-mediated METTL3/METTL14 transactivation were validated with chromatin immunoprecipitation polymerase chain reaction and dual-luciferase reporter system, and the role of m6A in TGF-ß1 upregulation was further verified in METTL3/METTL14-deficient KCs and myeloid lineage cell-specific METTL14 knockout mice. RESULTS: Serum lipopolysaccharide (LPS) concentration is increased in high-fat diet-induced NASH rats. TGF-ß1 upregulation is closely associated with METTL3/METTL14 upregulation and global m6A hypermethylation, in both NASH rat liver and LPS-activated KCs. LPS-responsive m6A peaks are identified on the 5' untranslated region (UTR) of TGF-ß1 messenger RNA (mRNA). NF-κB directly transactivates METTL3 and METTL14 genes. LPS-stimulated TGF-ß1 expression is abolished in METTL3/METTL14-deficient KCs and myeloid lineage cell-specific METTL14 knockout mice. Mutation of m6A sites on the 5'UTR of TGF-ß1 mRNA blocks LPS-induced increase of luciferase reporter activity. CONCLUSIONS: NF-κB acts as transcription factor to transactivate METTL3/METTL14 genes upon LPS challenge, leading to global RNA m6A hypermethylation. Increased m6A on the 5'UTR of TGF-ß1 mRNA results in m6A-dependent translation of TGF-ß1 mRNA in a cap-independent manner. We identify a novel role of m6A modification in TGF-ß1 upregulation, which helps to shed light on the molecular mechanism of NASH progression.


Subject(s)
Adenosine/analogs & derivatives , Kupffer Cells/metabolism , Methyltransferases/metabolism , Protein Biosynthesis , Transcriptional Activation , Transforming Growth Factor beta1/genetics , 5' Untranslated Regions , Adenosine/metabolism , Animals , Base Sequence , Diet, High-Fat , Disease Models, Animal , Gene Expression Regulation , Gene Knockdown Techniques , Liver Cirrhosis/etiology , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Methylation , Methyltransferases/deficiency , Models, Biological , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Transcription Factor RelA/metabolism , Transforming Growth Factor beta1/metabolism
12.
Cell Mol Gastroenterol Hepatol ; 12(2): 633-651, 2021.
Article in English | MEDLINE | ID: mdl-33848642

ABSTRACT

BACKGROUND & AIMS: N6-methyladenosine (m6A), the most prevalent and dynamic posttranscriptional methylation modification of mammalian mRNA, is involved in various biological processes, but its role in liver regeneration has not been characterized. METHODS: We first conducted transcriptome-wide m6A mRNA sequencing and characterized the expression pattern of m6A in regenerating mouse liver. Next, we generated hepatocyte-specific Mettl3- or Mettl14-deficient mice and investigated their role in liver regeneration. A series of biochemical experiments in vitro and in vivo was further performed to investigate potential mechanisms. RESULTS: We identified an overwhelming proportion of m6A-modified genes with initially up-regulated and subsequently down-regulated m6A levels as liver regeneration progressed. Loss of Mettl14 but not of Mettl3 resulted in markedly disrupted liver regeneration, and Mettl14-ablated hepatocytes were arrested in the G1 phase of the cell cycle. Most strikingly, the Mettl14-ablated regenerating liver exhibited extensive parenchymal necrosis. mRNA transcripts, such as Hsp90b1, Erp29, Stt3a, P4hb, and Lman1, encoding proteins involved in polypeptide processing and the endoplasmic reticulum (ER) stress response, were m6A-hypomethylated, and their mRNA and protein levels were subsequently decreased, resulting in unresolved ER stress, hepatocyte death, and inhibited proliferation. CONCLUSIONS: We demonstrate the essential role of Mettl14 in facilitating liver regeneration by modulating polypeptide-processing proteins in the ER in an m6A-dependent manner.


Subject(s)
Adenosine/analogs & derivatives , Endoplasmic Reticulum/metabolism , Homeostasis , Liver Regeneration , Methyltransferases/metabolism , Adenosine/metabolism , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cell Proliferation/drug effects , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/genetics , Gene Deletion , Hepatectomy , Hepatocytes/drug effects , Hepatocytes/pathology , Hepatocytes/ultrastructure , Homeostasis/drug effects , Homeostasis/genetics , Liver/metabolism , Liver/surgery , Liver Regeneration/drug effects , Liver Regeneration/genetics , Male , Methyltransferases/deficiency , Mice, Knockout , Necrosis , Organ Specificity/drug effects , Organ Specificity/genetics , Peptides/genetics , Peptides/metabolism , RNA Stability/drug effects , RNA Stability/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Taurochenodeoxycholic Acid/pharmacology , Transcriptome/genetics
13.
Sci Rep ; 11(1): 8119, 2021 04 14.
Article in English | MEDLINE | ID: mdl-33854089

ABSTRACT

Caffeine is the most consumed alkaloid stimulant in the world. It is synthesized through the activity of three known N-methyltransferase proteins. Here we are reporting on the 422-Mb chromosome-level assembly of the Coffea humblotiana genome, a wild and endangered, naturally caffeine-free, species from the Comoro archipelago. We predicted 32,874 genes and anchored 88.7% of the sequence onto the 11 chromosomes. Comparative analyses with the African Robusta coffee genome (C. canephora) revealed an extensive genome conservation, despite an estimated 11 million years of divergence and a broad diversity of genome sizes within the Coffea genus. In this genome, the absence of caffeine is likely due to the absence of the caffeine synthase gene which converts theobromine into caffeine through an illegitimate recombination mechanism. These findings pave the way for further characterization of caffeine-free species in the Coffea genus and will guide research towards naturally-decaffeinated coffee drinks for consumers.


Subject(s)
Coffea/genetics , Methyltransferases/genetics , Plant Proteins/genetics , Amino Acid Sequence , Caffeine/analysis , Chromosomes, Plant , Coffea/chemistry , Coffea/enzymology , Comoros , Comparative Genomic Hybridization , Evolution, Molecular , Methyltransferases/classification , Methyltransferases/deficiency , Phylogeny , Plant Leaves/chemistry , Plant Leaves/enzymology , Plant Leaves/genetics , Plant Proteins/classification , Plant Proteins/metabolism , Sequence Alignment , Sequence Analysis, RNA , Theobromine/analysis
14.
Nucleic Acids Res ; 49(2): e7, 2021 01 25.
Article in English | MEDLINE | ID: mdl-32710622

ABSTRACT

Traditional epitranscriptomics relies on capturing a single RNA modification by antibody or chemical treatment, combined with short-read sequencing to identify its transcriptomic location. This approach is labor-intensive and may introduce experimental artifacts. Direct sequencing of native RNA using Oxford Nanopore Technologies (ONT) can allow for directly detecting the RNA base modifications, although these modifications might appear as sequencing errors. The percent Error of Specific Bases (%ESB) was higher for native RNA than unmodified RNA, which enabled the detection of ribonucleotide modification sites. Based on the %ESB differences, we developed a bioinformatic tool, epitranscriptional landscape inferring from glitches of ONT signals (ELIGOS), that is based on various types of synthetic modified RNA and applied to rRNA and mRNA. ELIGOS is able to accurately predict known classes of RNA methylation sites (AUC > 0.93) in rRNAs from Escherichiacoli, yeast, and human cells, using either unmodified in vitro transcription RNA or a background error model, which mimics the systematic error of direct RNA sequencing as the reference. The well-known DRACH/RRACH motif was localized and identified, consistent with previous studies, using differential analysis of ELIGOS to study the impact of RNA m6A methyltransferase by comparing wild type and knockouts in yeast and mouse cells. Lastly, the DRACH motif could also be identified in the mRNA of three human cell lines. The mRNA modification identified by ELIGOS is at the level of individual base resolution. In summary, we have developed a bioinformatic software package to uncover native RNA modifications.


Subject(s)
Computational Biology/methods , High-Throughput Nucleotide Sequencing , RNA Processing, Post-Transcriptional , RNA-Seq , Scientific Experimental Error , Software , Adenine/analogs & derivatives , Adenine/analysis , Animals , Cell Line , Escherichia coli/genetics , Humans , Meiosis , Methyltransferases/deficiency , Methyltransferases/metabolism , Mice , Mice, Knockout , Nucleotide Motifs , RNA, Bacterial/genetics , RNA, Fungal/genetics , RNA, Messenger/genetics , RNA, Ribosomal/genetics , ROC Curve , Saccharomyces cerevisiae/genetics , Sequence Analysis, DNA , Templates, Genetic , Transcription, Genetic
15.
Pharmacogenomics ; 21(17): 1217-1226, 2020 11.
Article in English | MEDLINE | ID: mdl-33118454

ABSTRACT

Aim: To assess rare TPMT variants in patients carrying a deficient phenotype not predicted by the four more frequent genotypes (*2, *3A, *3B and *3C). Materials & methods: Next-generation sequencing of TPMT in 39 patients with a discordant genotype. Results: None of the variants identified explained the discordances assuming that they are of uncertain significance according to the Clinical Pharmacogenetics Implementation Consortium classification. Two unknown variants were detected and predicted to result in a splicing defect. We show that TPMT*16 and TMPT*21 are defective alleles, and TPMT*8 and TPMT*24 are associated with a normal activity. Conclusion: Whole-exon sequencing for rare TPMT mutations has a low diagnostic yield. A reassessment of the functional impact of rare variants of uncertain significance is a critical issue.


Subject(s)
Methyltransferases/deficiency , Methyltransferases/genetics , Alleles , Exons , Genetic Variation , Genotype , High-Throughput Nucleotide Sequencing , Humans , Introns/genetics , Mutation , Phenotype , Polymorphism, Genetic , Exome Sequencing
16.
Cell Rep ; 31(13): 107819, 2020 06 30.
Article in English | MEDLINE | ID: mdl-32610122

ABSTRACT

The RNA N6-methyladenosine (m6A) methylation is installed by the METTL3-METTL14 methyltransferase complex. This modification has critical regulatory roles in various biological processes. Here, we report that deletion of Mettl14 dramatically reduces mRNA m6A methylation in developing B cells and severely blocks B cell development in mice. Deletion of Mettl14 impairs interleukin-7 (IL-7)-induced pro-B cell proliferation and the large-pre-B-to-small-pre-B transition and causes dramatic abnormalities in gene expression programs important for B cell development. Suppression of a group of transcripts by cytoplasmic m6A reader YTHDF2 is critical to the IL-7-induced pro-B cell proliferation. In contrast, the block in the large-pre-B-to-small-pre-B transition is independent of YTHDF1 or YTHDF2 but is associated with a failure to properly upregulate key transcription factors regulating this transition. Our data highlight the important regulatory roles of the RNA m6A methylation and its reader proteins in early B cell development.


Subject(s)
Adenosine/analogs & derivatives , B-Lymphocytes/metabolism , RNA/metabolism , Adenosine/metabolism , Animals , Base Sequence , Cell Proliferation , Cell Size , Chromatin/metabolism , Immunoglobulin Heavy Chains/metabolism , Immunoglobulin Light Chains/metabolism , Interleukin-7/metabolism , Methylation , Methyltransferases/deficiency , Methyltransferases/metabolism , Mice, Knockout , Protein Binding , Protein Biosynthesis , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Transcription Factors/metabolism , Transcriptional Activation/genetics
17.
Nucleic Acids Res ; 48(11): 6251-6264, 2020 06 19.
Article in English | MEDLINE | ID: mdl-32406913

ABSTRACT

m6A is a prevalent internal modification in mRNAs and has been linked to the diverse effects on mRNA fate. To explore the landscape and evolution of human m6A, we generated 27 m6A methylomes across major adult tissues. These data reveal dynamic m6A methylation across tissue types, uncover both broadly or tissue-specifically methylated sites, and identify an unexpected enrichment of m6A methylation at non-canonical cleavage sites. A comparison of fetal and adult m6A methylomes reveals that m6A preferentially occupies CDS regions in fetal tissues. Moreover, the m6A sub-motifs vary between fetal and adult tissues or across tissue types. From the evolutionary perspective, we uncover that the selection pressure on m6A sites varies and depends on their genic locations. Unexpectedly, we found that ∼40% of the 3'UTR m6A sites are under negative selection, which is higher than the evolutionary constraint on miRNA binding sites, and much higher than that on A-to-I RNA modification. Moreover, the recently gained m6A sites in human populations are clearly under positive selection and associated with traits or diseases. Our work provides a resource of human m6A profile for future studies of m6A functions, and suggests a role of m6A modification in human evolutionary adaptation and disease susceptibility.


Subject(s)
Adenosine/analogs & derivatives , DNA Methylation , Evolution, Molecular , 3' Untranslated Regions , Adenosine/metabolism , Adult , Disease Susceptibility , Epigenome , Fetus/metabolism , Genetics, Population , HEK293 Cells , Humans , Methyltransferases/deficiency , Methyltransferases/genetics , Organ Specificity
18.
Nucleic Acids Res ; 48(11): 6157-6169, 2020 06 19.
Article in English | MEDLINE | ID: mdl-32392304

ABSTRACT

The TRM10 family of methyltransferases is responsible for the N1-methylation of purines at position 9 of tRNAs in Archaea and Eukarya. The human genome encodes three TRM10-type enzymes, of which only the mitochondrial TRMT10C was previously characterized in detail, whereas the functional significance of the two presumably nuclear enzymes TRMT10A and TRMT10B remained unexplained. Here we show that TRMT10A is m1G9-specific and methylates a subset of nuclear-encoded tRNAs, whilst TRMT10B is the first m1A9-specific tRNA methyltransferase found in eukaryotes and is responsible for the modification of a single nuclear-encoded tRNA. Furthermore, we show that the lack of G9 methylation causes a decrease in the steady-state levels of the initiator tRNAiMet-CAT and an alteration in its further post-transcriptional modification. Our work finally clarifies the function of TRMT10A and TRMT10B in vivo and provides evidence that the loss of TRMT10A affects the pool of cytosolic tRNAs required for protein synthesis.


Subject(s)
Methyltransferases/metabolism , tRNA Methyltransferases/metabolism , Base Sequence , Cell Line , Humans , Methylation , Methyltransferases/deficiency , Protein Biosynthesis , Purines/metabolism , RNA, Transfer/metabolism
19.
Tuberculosis (Edinb) ; 121: 101890, 2020 03.
Article in English | MEDLINE | ID: mdl-32279869

ABSTRACT

CpG motifs in DNA sequences are recognized by Toll-like receptor 9 and activate immune cells. Bacterial genomic DNA (gDNA) has modified cytosine bases (5-methylcytosine [5 mC]) and modified adenine bases (6-methyladenine [6 mA]). 5 mC inhibits immune activation by CpG DNA; however, it is unclear whether 6 mA inhibits immune activation by CpG DNA. Mycobacterium bovis BCG (BCG) has three adenine methyltransferases (MTases) that act on specific target sequences. In this study, we examined whether the 6 mA at the target sites of adenine MTases affected the immunostimulatory activity of CpG DNA. Our results showed that only 6 mA located at the target sequence of mamA, an adenine MTase from BCG, enhanced interleukin (IL)-12p40 production from murine bone marrow-derived macrophages (BMDMs) stimulated with CpG DNA. Enhancement of IL-12p40 production in BMDMs was also observed when BMDMs were stimulated with CpG DNA ligated to oligodeoxynucleotides (ODNs) harboring 6 mA. Accordingly, we then evaluated whether gDNA from adenine MTase-deficient BCG was less efficient with regard to stimulation of BMDMs. Indeed, gDNA from a mamA-deficient BCG had less ability to activate BMDMs than that from wild-type BCG. We concluded from these results that adenine methylation on ODNs and bacterial gDNA may enhance immune activity induced by CpG DNA.


Subject(s)
Adenine/analogs & derivatives , Adjuvants, Immunologic/pharmacology , DNA, Bacterial/immunology , Macrophage Activation/drug effects , Macrophages/drug effects , Methyltransferases/immunology , Mycobacterium bovis/immunology , Oligodeoxyribonucleotides/pharmacology , Toll-Like Receptor 9/agonists , Adenine/immunology , Animals , Cells, Cultured , DNA, Bacterial/genetics , Host-Pathogen Interactions , Interleukin-12 Subunit p40/metabolism , Macrophages/immunology , Macrophages/metabolism , Macrophages/microbiology , Methyltransferases/deficiency , Methyltransferases/genetics , Mice, Inbred C57BL , Mice, Knockout , Mycobacterium bovis/enzymology , Mycobacterium bovis/genetics , Signal Transduction , Toll-Like Receptor 9/genetics , Toll-Like Receptor 9/metabolism
20.
Int J Radiat Oncol Biol Phys ; 107(4): 779-792, 2020 07 15.
Article in English | MEDLINE | ID: mdl-32259569

ABSTRACT

PURPOSE: Acquired hematopoietic failure is commonly caused by therapeutic and accidental exposure of the bone marrow (BM) to toxic agents. Efficient recovery from BM failure is dictated not only by the intrinsic sensitivity and proliferation capacity of the hematopoietic stem and progenitor cells but also by the BM environment niche. Identification of genetic factors that improve recovery from hematopoietic failure is essential. Vertebrate SETD4 is a poorly characterized and putatively nonhistone methyltransferase. This study aims to identify the roles of SETD4 in BM recovery. METHODS AND MATERIALS: An inducible SETD4 knockout mouse model (Setd4flox/flox;Rosa26-CreERT2+) was used. Adult sex-matched littermates were treated with tamoxifen to induce Setd4 deletion or oil as the control. Tamoxifen-treated Setd4wt/wt;Rosa26-CreERT2+ mice were included as another control. Those mice were irradiated to induce hematopoietic syndrome and analyzed to identify the roles and mechanisms of Setd4 in of BM recovery. RESULTS: Loss of Setd4 in adult mice improved the survival of whole-body irradiation-induced BM failure. This was associated with improved recoveries of long-term and short-term hematopoietic stem cells (HSCs) and early progenitor cells. BM transplantation analyses surprisingly showed that the improved recovery was not due to radiation resistance of the Setd4-deficient HSCs but that Setd4-deficient HSCs were actually more sensitive to radiation. However, the Setd4-deficient mice were better recipients for allogeneic HSC transplantation. Furthermore, there was enhanced splenic erythropoiesis in Setd4-deficient mice. CONCLUSION: These findings not only revealed a previously unrecognized role of Setd4 as a unique modulator of hematopoiesis but also underscored the critical role of the BM niche in recovery from hematopoietic failure. Our study also implicated Setd4 as a potential target for therapeutic inhibition to improve the conditioning of the BM niche before allogeneic transplantation.


Subject(s)
Hematopoiesis/genetics , Hematopoiesis/radiation effects , Methyltransferases/deficiency , Methyltransferases/genetics , Animals , Bone Marrow Transplantation , Gene Knockout Techniques , Mice , Whole-Body Irradiation/adverse effects
SELECTION OF CITATIONS
SEARCH DETAIL
...